Introduction
The DEAD-box RNA helicase
DDX5 is overexpressed in multiple types of cancers in which its expression is necessary for cell proliferation and tumor growth (
1- Shin S.
- Rossow K.L.
- Grande J.P.
- Janknecht R.
Involvement of RNA helicases p68 and p72 in colon cancer.
,
2- Wortham N.C.
- Ahamed E.
- Nicol S.M.
- Thomas R.S.
- Periyasamy M.
- Jiang J.
- Ochocka A.M.
- Shousha S.
- Huson L.
- Bray S.E.
- Coombes R.C.
- Ali S.
- Fuller-Pace F.V.
The DEAD-box protein p72 regulates ERα-/oestrogen-dependent transcription and cell growth and is associated with improved survival in ERα-positive breast cancer.
,
3- Du C.
- Li D.Q.
- Li N.
- Chen L.
- Li S.S.
- Yang Y.
- Hou M.X.
- Xie M.J.
- Zheng Z.D.
DDX5 promotes gastric cancer cell proliferation in vitroin vivo through mTOR signaling pathway.
,
4- Clark E.L.
- Hadjimichael C.
- Temperley R.
- Barnard A.
- Fuller-Pace F.V.
- Robson C.N.
p68/Ddx5 supports β-catenin & RNAP II during androgen receptor mediated transcription in prostate cancer.
). Similar to other RNA helicases, DDX5 is involved in many steps of RNA metabolism, such as rRNA biogenesis, transcriptional regulation, alternative splicing, and microRNA processing (
5- Xing Z.
- Ma W.K.
- Tran E.J.
The DDX5/Dbp2 subfamily of DEAD-box RNA helicases.
). Our laboratory showed that DDX5 is an active RNA helicase
in vitro that is functionally conserved with its
Saccharomyces cerevisiae ortholog
DBP2 (
6- Xing Z.
- Wang S.
- Tran E.J.
Characterization of the mammalian DEAD-box protein DDX5 reveals functional conservation with S. cerevisiae ortholog Dbp2 in transcriptional control and glucose metabolism.
). We also found that deleting
DBP2 leads to global changes of mRNA secondary structures in
S. cerevisiae that correlate with altered transcriptional termination (
7- Lai Y.H.
- Choudhary K.
- Cloutier S.C.
- Xing Z.
- Aviran S.
- Tran E.J.
Genome-wide discovery of DEAD-box RNA helicase targets reveals RNA structural remodeling in transcription termination.
). Similarly, other groups have shown that DDX5 controls alternative splicing in a manner that may involve secondary structure remodeling (
8- Kar A.
- Fushimi K.
- Zhou X.
- Ray P.
- Shi C.
- Chen X.
- Liu Z.
- Chen S.
- Wu J.Y.
RNA helicase p68 (DDX5) regulates tau exon 10 splicing by modulating a stem-loop structure at the 5′ splice site.
,
9- Lee Y.J.
- Wang Q.
- Rio D.C.
Coordinate regulation of alternative pre-mRNA splicing events by the human RNA chaperone proteins hnRNPA1 and DDX5.
). This suggests that DDX5 and Dbp2 remodel RNA secondary structures in nascent RNA during transcription and pre-mRNA maturation steps, likely altering gene expression in the process.
In addition to RNA remodeling, DDX5 also acts as cofactor for oncogenic transcription factors in several cancer types (
2- Wortham N.C.
- Ahamed E.
- Nicol S.M.
- Thomas R.S.
- Periyasamy M.
- Jiang J.
- Ochocka A.M.
- Shousha S.
- Huson L.
- Bray S.E.
- Coombes R.C.
- Ali S.
- Fuller-Pace F.V.
The DEAD-box protein p72 regulates ERα-/oestrogen-dependent transcription and cell growth and is associated with improved survival in ERα-positive breast cancer.
,
10- Clark E.L.
- Coulson A.
- Dalgliesh C.
- Rajan P.
- Nicol S.M.
- Fleming S.
- Heer R.
- Gaughan L.
- Leung H.Y.
- Elliott D.J.
- Fuller-Pace F.V.
- Robson C.N.
The RNA helicase p68 is a novel androgen receptor coactivator involved in splicing and is overexpressed in prostate cancer.
).
DDX5 expression activates the oncogenic Wnt and mammalian target of rapamycin signaling pathways involved in cell-fate determination and cell growth (
3- Du C.
- Li D.Q.
- Li N.
- Chen L.
- Li S.S.
- Yang Y.
- Hou M.X.
- Xie M.J.
- Zheng Z.D.
DDX5 promotes gastric cancer cell proliferation in vitroin vivo through mTOR signaling pathway.
,
11P68 RNA helicase mediates PDGF-induced epithelial mesenchymal transition by displacing Axin from β-catenin.
,
12- Arun G.
- Akhade V.S.
- Donakonda S.
- Rao M.R.
mrhl RNA, a long noncoding RNA, negatively regulates Wnt signaling through its protein partner Ddx5/p68 in mouse spermatogonial cells.
). Interestingly, depletion of
DDX5 in mammalian cells or deletion of
DBP2 in
S. cerevisiae lead to differential expression of metabolic genes (
13- Mazurek A.
- Park Y.
- Miething C.
- Wilkinson J.E.
- Gillis J.
- Lowe S.W.
- Vakoc C.R.
- Stillman B.
Acquired dependence of acute myeloid leukemia on the DEAD-box RNA helicase DDX5.
,
14- Beck Z.T.
- Cloutier S.C.
- Schipma M.J.
- Petell C.J.
- Ma W.K.
- Tran E.J.
Regulation of glucose-dependent gene expression by the RNA helicase Dbp2 in Saccharomyces cerevisiae.
) and alteration of glycolysis and mitochondrial respiration activity (
6- Xing Z.
- Wang S.
- Tran E.J.
Characterization of the mammalian DEAD-box protein DDX5 reveals functional conservation with S. cerevisiae ortholog Dbp2 in transcriptional control and glucose metabolism.
,
15- Wang S.
- Xing Z.
- Pascuzzi P.E.
- Tran E.J.
Metabolic adaptation to nutrients involves coregulation of gene expression by the RNA helicase Dbp2 and the Cyc8 corepressor in Saccharomyces cerevisiae.
). Moreover, a
DDX5 S480A polymorphism has been connected to metabolic syndrome in humans, which is typified by abnormalities in primary metabolic pathways (
16- Guo J.
- Hong F.
- Loke J.
- Yea S.
- Lim C.L.
- Lee U.
- Mann D.A.
- Walsh M.J.
- Sninsky J.J.
- Friedman S.L.
A DDX5 S480A polymorphism is associated with increased transcription of fibrogenic genes in hepatic stellate cells.
). This suggests that the DDX5 family is involved in energy metabolism, a process that is misregulated in many cancers (
17- Pavlova N.N.
- Thompson C.B.
The emerging hallmarks of cancer metabolism.
).
Small cell lung cancer (SCLC) accounts for 15% of lung cancer cases and has a 5-year survival rate of less than 10% (
18- Gazdar A.F.
- Bunn P.A.
- Minna J.D.
Small-cell lung cancer: what we know, what we need to know and the path forward.
). SCLC is an aggressive disease because of its fast growth rate and early development of metastases. This cancer shows positive initial response to chemotherapies but develops resistance rapidly, resulting in relapse within 2 years. SCLC originates from neuroendocrine cells, with the classic subtype of SCLC defined by expression of the neuroendocrine-specific transcriptional factor achaete-scute homolog 1, a lineage-specific oncogene (
18- Gazdar A.F.
- Bunn P.A.
- Minna J.D.
Small-cell lung cancer: what we know, what we need to know and the path forward.
,
19- Asai N.
- Ohkuni Y.
- Kaneko N.
- Yamaguchi E.
- Kubo A.
Relapsed small cell lung cancer: treatment options and latest developments.
). SCLC invariably inactivates the tumor suppressor genes
TP53 and RB transcriptional corepressor 1. Factors such as the
MYC family of oncogenes, the apoptosis regulator Bcl-2, and the tetraspanin cell-surface family member CD151 have been correlated with SCLC (
20Targets in small cell lung cancer.
). However, SCLC still remains largely uncharacterized, and there are currently no effective pharmacological strategies to prevent recurrence of the disease.
Despite diverse genetic landscapes, a common theme of cellular transformation is up-regulation of metabolic pathways to support rapid growth and biomass production (
21- DeBerardinis R.J.
- Chandel N.S.
Fundamentals of cancer metabolism.
). The observation by Otto Warburg in the 1920's that cancer cells continue to convert glucose to lactate through lactic acid fermentation, despite the availability of oxygen to support respiration, has now been observed in multiple cancer cell lines and within solid tumors (
22- Koppenol W.H.
- Bounds P.L.
- Dang C.V.
Otto Warburg's contributions to current concepts of cancer metabolism.
). It is now established that this type of “reprogramed” metabolism is not used for ATP production but rather to produce glycolytic intermediates that form building blocks for rapid cellular growth. Importantly, cancer cells still generate ATP through mitochondrial respiration to fuel biosynthesis (
21- DeBerardinis R.J.
- Chandel N.S.
Fundamentals of cancer metabolism.
). Thus, mitochondrial defects can be a barrier for malignancy, suggesting the necessity of mitochondrial functions in carcinogenesis. Herein, we show that
DDX5 depletion leads to growth defects and global changes of gene expression in drug-resistant SCLC cells. Moreover, we find that DDX5 is necessary for mitochondrial function and production of the TCA cycle intermediate succinate. This suggests that DDX5 may be a novel drug target for small cell lung cancer in the future.
Discussion
DEAD-box RNA helicases are the largest class of enzymes in the RNA helicase family, acting as nonprocessive, ATP-dependent RNA-binding proteins, whose activity can be coupled to remodeling of secondary structure and/or RNA–protein complexes (
32- Jarmoskaite I.
- Russell R.
DEAD-box proteins as RNA helicases and chaperones.
). Somewhat paradoxically, up-regulation of members of the DEAD-box protein family, including
DDX1,
DDX2,
DDX3, and
DDX5, has been linked to cellular transformation and correlates with multiple cancer types including breast, colon, bone, and prostate (
33The DEAD box proteins DDX5 (p68) and DDX17 (p72): multi-tasking transcriptional regulators.
,
34- Mohibi S.
- Chen X.
- Zhang J.
Cancer the'RBP'eutics–RNA-binding proteins as therapeutic targets for cancer.
). Herein, we assessed the role of DEAD-box helicase DDX5 in drug-resistant SCLC, a highly aggressive, metastatic cancer with a high mortality rate. Our results show that DDX5 is required for anchorage-independent growth and normal respiration of SCLC cells, consistent with the notion that mitochondrial functions are necessary for carcinogenesis (
35- Joshi S.
- Tolkunov D.
- Aviv H.
- Hakimi A.A.
- Yao M.
- Hsieh J.J.
- Ganesan S.
- Chan C.S.
- White E.
The genomic landscape of renal oncocytoma identifies a metabolic barrier to tumorigenesis.
,
36- Weinberg F.
- Hamanaka R.
- Wheaton W.W.
- Weinberg S.
- Joseph J.
- Lopez M.
- Kalyanaraman B.
- Mutlu G.M.
- Budinger G.R.
- Chandel N.S.
Mitochondrial metabolism and ROS generation are essential for Kras-mediated tumorigenicity.
). Moreover, we find that
DDX5 expression is necessary for accumulation of succinate, an oncometabolite whose up-regulation correlates with chemoresistance (
37Cancer metabolism and drug resistance.
), suggesting that up-regulation of
DDX5 may be part of the underlying mechanism for drug resistance in SCLC.
DDX5 was one of the first members of the DEAD-box helicase family to be enzymatically characterized and, in the 30 years since, has now been implicated in multiple transcriptional and cotranscriptional processes suggestive of a role as an RNA chaperone for nascent transcripts (
5- Xing Z.
- Ma W.K.
- Tran E.J.
The DDX5/Dbp2 subfamily of DEAD-box RNA helicases.
). This raises the question of how a general chaperone that acts in a wide array of gene expression steps could promote uncontrolled cell growth. Although we do not know the precise molecular basis of this activity, two possibilities exist for a putative mechanism for DDX5 in supporting rapid cellular proliferation. First, a biochemically distinct pool of DDX5 may exist
in vivo that regulates transcription of specific genes directly. DDX5 is both polyubiquitinated and phosphorylated, modifications that appear to correlate with distinct activities. Specifically, phosphorylation of Tyr-593 at the C terminus of DDX5 promotes cellular proliferation and expression of genes involved in cell growth such as
c-MYC and
Cyclin D1 (
11P68 RNA helicase mediates PDGF-induced epithelial mesenchymal transition by displacing Axin from β-catenin.
,
23DEAD box RNA helicase functions in cancer.
). Interestingly, DDX5 was recently shown to unwind a transcriptionally repressive G-quadraplex in the
c-MYC promoter, illustrating an unexpected, DNA-dependent activity for this enzyme (
38- Wu G.
- Xing Z.
- Tran E.J.
- Yang D.
DDX5 helicase resolves G-quadruplex and is involved in MYC gene transcriptional activation.
). Direct regulation of transcriptional activity is supported by ChIP profiles, which show enrichment of DDX5 at promoters of genes required for mitochondrial function (
Fig. 3,
B and
C). Second, the secondary structure of a given pre-mRNA may determine the requirement for DDX5. In support of this, studies from our laboratory have shown that the budding yeast counterpart of DDX5, termed Dbp2, promotes cellular metabolism and expression of genes involved in energy homeostasis (
15- Wang S.
- Xing Z.
- Pascuzzi P.E.
- Tran E.J.
Metabolic adaptation to nutrients involves coregulation of gene expression by the RNA helicase Dbp2 and the Cyc8 corepressor in Saccharomyces cerevisiae.
). Interestingly, Dbp2 appears to regulate gene expression at the level of transcription termination by remodeling mRNA structure, suggesting that the underlying secondary structure of RNA may confer Dbp2 dependence to the expression of specific genes (
7- Lai Y.H.
- Choudhary K.
- Cloutier S.C.
- Xing Z.
- Aviran S.
- Tran E.J.
Genome-wide discovery of DEAD-box RNA helicase targets reveals RNA structural remodeling in transcription termination.
). DDX5 is functionally equivalent to
S. cerevisiae Dbp2 (
6- Xing Z.
- Wang S.
- Tran E.J.
Characterization of the mammalian DEAD-box protein DDX5 reveals functional conservation with S. cerevisiae ortholog Dbp2 in transcriptional control and glucose metabolism.
,
39- Iggo R.D.
- Jamieson D.J.
- MacNeill S.A.
- Southgate J.
- McPheat J.
- Lane D.P.
p68 RNA helicase: identification of a nucleolar form and cloning of related genes containing a conserved intron in yeasts.
), suggesting that DDX5 may function similarly, depending primarily on signatures that reside in the nascent gene transcript itself. A recent study showing that DDX5-binding sites are in close proximity to structured regions near exons suggests that a similar mechanism may be involved in alternative splicing (
9- Lee Y.J.
- Wang Q.
- Rio D.C.
Coordinate regulation of alternative pre-mRNA splicing events by the human RNA chaperone proteins hnRNPA1 and DDX5.
), although is unknown whether DDX5 functions in termination in mammalian cells. Future studies are necessary to determine the precise mechanism by which DDX5 promotes respiration.
Our studies show that loss of DDX5 in SCLC cells impairs mitochondrial function, a defect that is consistent with up-regulation of mtDNA and, likely, accumulation of defective mitochondria (
26- Shapovalov Y.
- Hoffman D.
- Zuch D.
- de Mesy Bentley K.L.
- Eliseev R.A.
Mitochondrial dysfunction in cancer cells due to aberrant mitochondrial replication.
). This is in contrast to our previous results in mouse liver cells whereby
DDX5 knockdown leads to down-regulation of glycolysis and up-regulation of respiration (
6- Xing Z.
- Wang S.
- Tran E.J.
Characterization of the mammalian DEAD-box protein DDX5 reveals functional conservation with S. cerevisiae ortholog Dbp2 in transcriptional control and glucose metabolism.
).
DDX5 expression is below detection by Western blotting in normal lung epithelial HBEC cells and tested NSCLC cells (
Fig. 1A), suggesting that DDX5 is not required for the growth of these cell lines. Similarly, ablation of
DDX5 expression in mESC or human MCF-7 cells does not impact cell proliferation, and knockdown in HeLa cells (to 5% of control siRNA) only slightly alters the proliferation rate (
2- Wortham N.C.
- Ahamed E.
- Nicol S.M.
- Thomas R.S.
- Periyasamy M.
- Jiang J.
- Ochocka A.M.
- Shousha S.
- Huson L.
- Bray S.E.
- Coombes R.C.
- Ali S.
- Fuller-Pace F.V.
The DEAD-box protein p72 regulates ERα-/oestrogen-dependent transcription and cell growth and is associated with improved survival in ERα-positive breast cancer.
,
40- Li H.
- Lai P.
- Jia J.
- Song Y.
- Xia Q.
- Huang K.
- He N.
- Ping W.
- Chen J.
- Yang Z.
- Li J.
- Yao M.
- Dong X.
- Zhao J.
- Hou C.
- et al.
RNA helicase DDX5 inhibits reprogramming to pluripotency by miRNA-based repression of RYBP and its PRC1-dependent and -independent functions.
,
41- Jalal C.
- Uhlmann-Schiffler H.
- Stahl H.
Redundant role of DEAD box proteins p68 (Ddx5) and p72/p82 (Ddx17) in ribosome biogenesis and cell proliferation.
).
DDX5 expression is developmentally regulated and is not detectable in mouse embryos until embryonic day 11.5 (
42- Stevenson R.J.
- Hamilton S.J.
- MacCallum D.E.
- Hall P.A.
- Fuller-Pace F.V.
Expression of the “dead box” RNA helicase p68 is developmentally and growth regulated and correlates with organ differentiation/maturation in the fetus.
). Moreover, DDX5 is dispensable for normal hematopoiesis and tissue functions in adult mice (
13- Mazurek A.
- Park Y.
- Miething C.
- Wilkinson J.E.
- Gillis J.
- Lowe S.W.
- Vakoc C.R.
- Stillman B.
Acquired dependence of acute myeloid leukemia on the DEAD-box RNA helicase DDX5.
) and for the function of mature adipocytes, despite being required to initiate adipogenesis (
43- Ramanathan N.
- Lim N.
- Stewart C.L.
DDX5/p68 RNA helicase expression is essential for initiating adipogenesis.
). These data suggest that DDX5 is dispensable in many, fully differentiated cell types, and we reason that the differences are likely due to cell type–/tissue-specific metabolism (
i.e. mouse hepatocyte
versus transformed human lung cell). For example, the hepatocyte-specific glucose transporter 2 and glucokinase regulator encode proteins that mediate glucose import and regulate glucose metabolism through inhibiting glucokinase, respectively (
44- Karim S.
- Adams D.H.
- Lalor P.F.
Hepatic expression and cellular distribution of the glucose transporter family.
,
45- Raimondo A.
- Rees M.G.
- Gloyn A.L.
Glucokinase regulatory protein: complexity at the crossroads of triglyceride and glucose metabolism.
). We also observed down-regulation of glycolysis in
S. cerevisiae cells upon deletion of
DBP2 (
15- Wang S.
- Xing Z.
- Pascuzzi P.E.
- Tran E.J.
Metabolic adaptation to nutrients involves coregulation of gene expression by the RNA helicase Dbp2 and the Cyc8 corepressor in Saccharomyces cerevisiae.
), consistent with the fact that budding yeast also rely predominantly on glycolysis over respiration when glucose is abundant (
46- Diaz-Ruiz R.
- Rigoulet M.
- Devin A.
The Warburg and Crabtree effects: on the origin of cancer cell energy metabolism and of yeast glucose repression.
).
In addition to mitochondrial dysfunction, we also observed a striking decrease in succinate accumulation in SCLC cells upon
DDX5 knockdown. This decrease in necessary metabolites may account for loss of respiration and slowed growth because succinate is necessary for both oxidative phosphorylation and the TCA cycle (
Fig. 5B). Interestingly, succinate has been described as an oncometabolite, along with fumarate and 2-hydroxyglutarate, that functions both in primary metabolism but also facilitates epigenetic changes by inhibiting α-ketoglutarate dioxygenases (
47- Dalla Pozza E.
- Dando I.
- Pacchiana R.
- Liboi E.
- Scupoli M.T.
- Donadelli M.
- Palmieri M.
Regulation of succinate dehydrogenase and role of succinate in cancer.
). Tumors with succinate dehydrogenase mutations that cause succinate accumulation are highly metastatic and aggressive, displaying decreased expression of genes needed for cellular differentiation and/or stabilizing HIF1α, which promotes cell growth and survival (
48- Selak M.A.
- Armour S.M.
- MacKenzie E.D.
- Boulahbel H.
- Watson D.G.
- Mansfield K.D.
- Pan Y.
- Simon M.C.
- Thompson C.B.
- Gottlieb E.
Succinate links TCA cycle dysfunction to oncogenesis by inhibiting HIF-α prolyl hydroxylase.
,
49Succinate: an initiator in tumorigenesis and progression.
). One possible explanation for the accumulation of succinate in SCLC cells involves tumor necrosis factor receptor–associated protein 1 (TRAP1), a member of the heat shock protein 90 family shown to be overexpressed in SCLC and associated with drug resistance in various cancers (
47- Dalla Pozza E.
- Dando I.
- Pacchiana R.
- Liboi E.
- Scupoli M.T.
- Donadelli M.
- Palmieri M.
Regulation of succinate dehydrogenase and role of succinate in cancer.
,
50- Lee J.H.
- Kang K.W.
- Kim J.E.
- Hwang S.W.
- Park J.H.
- Kim S.H.
- Ji J.H.
- Kim T.G.
- Nam H.Y.
- Roh M.S.
- Lee E.H.
- Park M.I.
- Kim M.S.
- Lee H.W.
Differential expression of heat shock protein 90 isoforms in small cell lung cancer.
,
51- Matassa D.S.
- Agliarulo I.
- Avolio R.
- Landriscina M.
- Esposito F.
TRAP1 regulation of cancer metabolism: dual role as oncogene or tumor suppressor.
). TRAP1 down-regulates SDH activity and induces succinate accumulation and HIF1α stabilization (
52- Sciacovelli M.
- Guzzo G.
- Morello V.
- Frezza C.
- Zheng L.
- Nannini N.
- Calabrese F.
- Laudiero G.
- Esposito F.
- Landriscina M.
- Defilippi P.
- Bernardi P.
- Rasola A.
The mitochondrial chaperone TRAP1 promotes neoplastic growth by inhibiting succinate dehydrogenase.
). Regulation of
TRAP1 expression by DDX5 could then help explain the decrease of succinate accumulation and proliferation observed upon
DDX5 knockdown. Future studies are needed to determine both how DDX5 promotes succinate accumulation and whether reduction of succinate is both necessary and sufficient to inhibit SCLC growth and invasion.
SCLC is a highly aggressive lung cancer with a median survival of 9 months that encompasses ∼15% of all lung cancer cases worldwide (
RRID:SCR_018570). There are currently no effective treatment options or early detection methods to date, making SCLC a major health challenge around the world. Studying SCLC has been a challenge for the community because of the dearth of patient samples, however, with characterization of cell lines and limited tissue samples providing the majority of mechanistic insight. Understanding the underlying mechanisms of cellular transformation and establishment of chemoresistance in SCLC requires defining key pathways that support this aggressive cancer with the goal of developing future targeted chemotherapies.
Experimental procedures
Plasmids and shRNAs
Plasmids and primers used for cloning and expression can be found in
Tables S1 and S2, respectively.
DDX5 cDNA (Gene ID: 1655, Dharmacon: 3528578) was subcloned into the PAcGFP1-Hyg-C1 vector to generate pAcGFP-Hyg-DDX5, using PCR primers HindIII DDX5 forward and SalI DDX5 reverse. The
shDDX5 (sh2)–resistant pGFP–DDX5 was then generated using site-directed mutagenesis with primers
shDDX5-resistant DDX5 forward and reverse.
The SMARTvector-inducible lentiviral shRNAs (Dharmacon) were used for depleting DDX5. A nonsilencing shRNA was used as a control (VSC11651), and two shRNAs were used to target DDX5 (V3SH11252): shDDX5 (ATCCAATCCACTTAGAGCA) and shDDX5-2 (TTAGAACCCAGTCACGCTC). We generated the lentiviral particles for transduction of the shRNAs using the Trans-lentiviral packaging system (Dharmacon, TLP5917).
Cell culture, transfection, and lentivirus transduction
H69 and H69AR cells were obtained from the American Type Culture Collection and cultured as instructed. The HBEC-3KT (HBEC) cell line was a gift from Dr. Andrea Kasinski (Purdue University) and was cultured with Keratinocyte-SFM medium (Gibco, 17005042). Transfection was performed with Lipofectamine 2000 reagent (Invitrogen, 11668027). For lentivirus transduction (TU), a multiplicity of infection of <0.3 TU/cell was used to prevent multiple genome insertion events. Briefly, the cells were mixed with 6 µg/ml Polybrene and virus in the absence of serum (fetal bovine serum). 10% or 20% fetal bovine serum was added to the mixture after a 6-h incubation in a humidified CO2 incubator for H69 or H69AR cells, respectively. After 24 h, the virus-containing medium was replaced with fresh medium. For H69 cells, the cell–virus mixture was centrifuged at 300 × g for 1 h at 25 °C before being put into the incubator.
Western blotting
The cells were lysed using 1× lysis buffer (Cell Signaling, 9803) with the addition of protease inhibitor (Roche, 11873580001). Protein levels were analyzed using the following antibodies: anti-DDX5 (Millipore, 05-850), anti–β-actin (Sigma, A5441), anti-PARP (Cell Signaling, 9532), anti–N-Myc (Abiocode, R1281-2), anti–c-Myc (Cell Signaling, D84C12), anti-Glut1 (Santa Cruz, SC377228), anti-CD151 (Santa Cruz, SC271216), and anti-Bcl-2 (Santa Cruz, SC7382).
Growth analysis
To measure growth in normal culture conditions, H69AR cells were treated with 1 µg/ml doxycycline for 3 days and subsequently seeded at 400 cells/well in 96-well plates (Corning, 3603). Live cells were labeled with DNA-binding fluorescent dye on indicated days using the CyQUANT® direct cell proliferation kit (Thermo Fisher, C35011) during an 8-day period. We used the relative fluorescence to infer the cell count. Background fluorescence from unlabeled cells was subtracted from the relative fluorescence. Doubling times were calculated using GraphPad Prism.
Soft agar assays were conducted as described (
53- Borowicz S.
- Van Scoyk M.
- Avasarala S.
- Karuppusamy Rathinam M.K.
- Tauler J.
- Bikkavilli R.K.
- Winn R.A.
The soft agar colony formation assay.
). H69 (10,000 cells/well) or H69AR (5000 cells/well) were treated with 1 µg/ml doxycycline for 3 days and then seeded in 6-well plates. Assays with H69 cells were incubated for 30 days, and assays with H69AR cells were incubated for 21 days.
RNA-Seq and data analysis
Three biological replicates of the H69AR cells stably transfected with shCtr or shDDX5 were used for RNA-Seq. Total RNA was extracted using TRIzol (Thermo Fisher, 15596026), treated with DNase (Thermo Fisher, AM2238), and then subjected to library preparation using the TruSeq® stranded total RNA library prep kit (Illumina, 20020596). Unique dual index adapters (TruSeq RNA UD Indexes Illumina, 20022371) were substituted for the single index adapters. Libraries were clustered on Illumina NovaSeq S4 (300 cycle) cassettes for paired-end, 150-bp read sequencing.
Data quality control (minimum Phred score, 30; minimum read length, 50 bp) was performed using the TrimGalore toolkit (version 0.4.4) (
RRID:SCR_011847). Quality trimmed reads were mapped to human reference genome (GRCh38) downloaded from the Ensembl database, using the STAR aligner (version 2.5.4b) (
54- Dobin A.
- Davis C.A.
- Schlesinger F.
- Drenkow J.
- Zaleski C.
- Jha S.
- Batut P.
- Chaisson M.
- Gingeras T.R.
STAR: ultrafast universal RNA-Seq aligner.
). Reads aligned to each gene feature were counted via HTSeq package, followed by differential expression analysis using DESeq2 and edgeR methods (
55- Love M.I.
- Huber W.
- Anders S.
Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2.
,
56- Robinson M.D.
- McCarthy D.J.
- Smyth G.K.
edgeR: a Bioconductor package for differential expression analysis of digital gene expression data.
). Genes with FDR ≤ 0.05 and differential expression by both methods were denoted as significant. Functional and pathway analysis was performed with 6727 significant DEGs using IPA (
57- Krämer A.
- Green J.
- Pollard Jr., J.
- Tugendreich S.
Causal analysis approaches in Ingenuity Pathway Analysis.
). Pathways with IPA assigned
p values of <0.05 were denoted as significant, and pathway direction was determined by assigned
z scores.
DDX5 ChIP-Seq reanalysis
The DDX5 ChIP-Seq data were derived from published data sets (
25- Yao H.
- Brick K.
- Evrard Y.
- Xiao T.
- Camerini-Otero R.D.
- Felsenfeld G.
Mediation of CTCF transcriptional insulation by DEAD-box RNA-binding protein p68 and steroid receptor RNA activator SRA.
). Raw reads of DDX5 ChIP and input were downloaded from Gene Expression Omnibus with accession number GSE24126. Adaptor sequences were removed using Trimmomatic (version 0.36). Reads were aligned to the human genome (GRCh38) using bowtie (version 1.2.2). Duplications are marked and removed using Picard tools (version 1.9). The peaks were determined by MACS2 narrow peak calling (version 2.1.2) with FDR (
q value) cutoff of 0.05. The figure of peaks and signal intensity was generated through Gviz R package (version 1.28.1).
RT-qPCR
Total RNA was extracted and subjected to reverse transcription using the QuantiTect kit (Qiagen, 205310). qPCR was conducted using the SYBR green master mix (Applied Biosystems, 4309155) to analyze the expression levels of specific mature mRNAs using primer pairs spanning exon-junctions (
Table S3). The relative expression level is calculated using the ΔΔ
Cq method using glyceraldehyde-3-phosphate dehydrogenase as the reference gene.
EdU incorporation
H69AR cells were seeded in 96-well plates at 40000 cells/well 24 h prior to the experiment. The cells were incubated with 10 μm EdU for 4 h, fixed with 3.7% formaldehyde, and permeabilized. The relative amount of incorporated EdU was then quantified using the Click-iTTM EdU Alexa FluorTM 647 HCS assay (Invitrogen, C10356).
Metabolic analysis
To measure extracellular flux, 20,000 cells/well of H69AR cells or 40,000 cells/well of HBEC cells were seeded in 24-well plates. H69AR cells were treated with 1 µg/ml doxycycline for 3 days before seeding. The respiration profiles of these cells were measured using the Seahorse XFe24 metabolic flux analyzer according to the Mito-stress test kit (Agilent, 103015-100) protocols. Basal glycolysis rates were measured at the same time, but before adding any drug from the kit. Data analysis was performed using the Seahorse report generator (Agilent). For assays with metformin treatment, metformin was added to the cells 24 h prior to the experiment. H69 cells could not be analyzed because they are nonadherent and form aggregates.
For lactate quantification, H69AR cells were treated with 1 µg/ml doxycycline for 3 days and then seeded in 6-well plates. After 40 h, the cells were washed using PBS and lysed. Intracellular lactate concentrations were measured according to the lactate assay kit (Sigma, MAK064).
Mitochondrial morphology characterization
H69AR cells stably transfected with doxycycline-inducible shRNAs targeting DDX5 or with a nontargeting control were treated with 1 µg/ml doxycycline for 48 h to induce shRNA expression. Both cell lines were seeded at 0.5 × 106 cells/well onto polylysine-treated coverslips in a 6-well plate and left to grow overnight with 1 µg/ml doxycycline. The cells were then treated with 500 nm MitoTracker Deep Red (Thermo Fisher Scientific, M22426) for mitochondrial visualization, according to the manual. The cells were fixed using 0.6% formaldehyde, and DNA was stained with 300 nm DAPI to serve as a nuclear marker. The cells were visualized using a Leica DM6 microscope with a 40× objective.
DAPI-stained nuclei were measured to compare cell size, because typical nuclei boundaries are more easily defined than the irregularly shaped outer boundaries of the cell. Leica LAS X software was utilized for nuclei measurement.
TCA metabolite profiling
Sample preparation
H69AR cells were grown in the presence of 1 µg/ml doxycycline for 3 days, and 100 mg of cells were pelleted and washed. A Bligh–Dyer extraction, a standard procedure used to isolate total lipid fractions from biological samples (
58A rapid method of total lipid extraction and purification.
), was performed to lyse the cells and extract the TCA cycle intermediates. 300 µl of methanol and 150 µl of water was added to the microcentrifuge tube, followed by sonication for 5 min. 300 µl of chloroform and 150 µl water was added. The samples were vortexed and then centrifuged. The top layer was lyophilized and reconstituted in 100 µl of 50% acetonitrile and 50% water prior to HPLC–MS/MS analysis.
HPLC–MS/MS
TCA cycle intermediates were analyzed as described (
29- Al Kadhi O.
- Melchini A.
- Mithen R.
- Saha S.
Development of a LC-MS/MS method for the simultaneous detection of tricarboxylic acid cycle intermediates in a range of biological matrices.
), using an Agilent 1290 Infinity II HPLC and 6470 triple quadrupole mass spectrometer. An Agilent HILIC-Z (2.1 × 150 mm, 2.7 μm) column was used at a flow rate of 0.3 ml/min. Mobile phase A was 90% acetonitrile, 5% isopropanol, and 5% 200 m
m ammonium acetate in water at pH 9. Mobile phase B was 90% water, 5% isopropanol, and 5% 200 m
m ammonium acetate in water at pH 9. A linear gradient elution was used as follows: initial conditions 5% B; 0–5 min: isocratic at 5% B; 5–15 min: gradient to 100% B; 15–16 min: isocratic at 100% B. Column re-equilibration was 16–17 min: gradient to 5% B; 17–20 min: isocratic at 5% B. Effluent was transferred to the mass spectrometer using negative electrospray ionization. Identification was based on retention time of authentic standards (pure TCA metabolites) and multiple reaction monitoring.
Article info
Publication history
Published online: May 06, 2020
Received in revised form:
April 23,
2020
Received:
January 9,
2020
Edited by John M. Denu
Footnotes
This article contains supporting information.
Author contributions—Z. X. and E. J. T. conceptualization; Z. X. and M. P. R. data curation; Z. X. validation; Z. X. and M. P. R. investigation; Z. X. and S. M. U. methodology; Z. X., M. P. R., and S. M. U. writing-original draft; M. P. R. and E. J. T. writing-review and editing; S. M. U. formal analysis; E. J. T. supervision; E. J. T. funding acquisition.
Funding and additional information—This work was supported by funds from the Department of Biochemistry and Purdue University Center for Cancer Research through National Institutes of Health Grant P30 CA023168 and by a kind gift from Mary Slevin in honor of her parents. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.
Conflict of interest—The authors declare that they have no conflicts of interest with the contents of this article.
Present address for Zheng Xing: University of Texas Southwestern Medical Center, Dallas, Texas, USA.
Abbreviations—The abbreviations used are:
ChIP-Seq
ChIP sequencing
SCLCsmall cell lung cancer
NSCLCnon–small cell lung cancer
HBECHBEC-3KT
shRNAshort hairpin RNA
shDDX5DDX5-targeting shRNA
shCtrnontargeting shRNA
siRNAsmall interfering RNA
EdU5-ethynyl-2′-deoxyuridine
IPAIngenuity Pathway Analysis
OxPhosoxidative phosphorylation
TSStranscription start site
DEGdifferentially expressed gene
mtDNAmitochondrial DNA
TRAP1tumor necrosis factor receptor–associated protein 1
c-MycMyc proto-oncogene protein
N-Mycneuroblastoma Myc
PARPpoly(ADP-ribose) polymerase
GFPgreen fluorescent protein
RNA-SeqRNA sequencing
FDRfalse discovery rate
qPCRquantitative PCR
DAPI4′,6′-diamino-2-phenylindole
TCAtricarboxylic acid
.Copyright
© 2020 Xing et al.